throbber
Current Medicinal Chemistry, 2003, 10, 2471-2483
`2471
`Glucagon-Like Peptide-1 Synthetic Analogs: New Therapeutic Agents for
`Use in the Treatment of Diabetes Mellitus
`
`George G. Holz* and Oleg G. Chepurny
`
`New York University School of Medicine, Department of Physiology and Neuroscience, New York, NY, USA
`
`Abstract: Glucagon-like peptide-1-(7-36)-amide (GLP-1) is a potent blood glucose-lowering hormone now
`under investigation for use as a therapeutic agent in the treatment of type 2 (adult onset) diabetes mellitus.
`GLP-1 binds with high affinity to G protein-coupled receptors (GPCRs) located on pancreatic β-cells, and it
`exerts insulinotropic actions that include the stimulation of insulin gene transcription, insulin biosynthesis,
`and insulin secretion. The beneficial therapeutic action of GLP-1 also includes its ability to act as a growth
`factor, stimulating formation of new pancreatic islets (neogenesis) while slowing β-cell death (apoptosis). GLP-
`1 belongs to a large family of structurally-related hormones and neuropeptides that include glucagon, secretin,
`GIP, PACAP, and VIP. Biosynthesis of GLP-1 occurs in the enteroendocrine L-cells of the distal intestine, and
`the release of GLP-1 into the systemic circulation accompanies ingestion of a meal. Although GLP-1 is
`inactivated rapidly by dipeptidyl peptidase IV (DDP-IV), synthetic analogs of GLP-1 exist, and efforts have
`been directed at engineering these peptides so that they are resistant to enzymatic hydrolysis. Additional
`modifications of GLP-1 incorporate fatty acylation and drug affinity complex (DAC) technology to improve
`serum albumin binding, thereby slowing renal clearance of the peptides. NN2211, LY315902, LY307161, and
`CJC-1131 are GLP-1 synthetic analogs that reproduce many of the biological actions of GLP-1, but with a
`prolonged duration of action. AC2993 (Exendin-4) is a naturally occurring peptide isolated from the lizard
`Heloderma, and it acts as a high affinity agonist at the GLP-1 receptor. This review summarizes structural
`features and signal transduction properties of GLP-1 and its cognate β-cell GPCR. The usefulness of synthetic
`GLP-1 analogs as blood glucose-lowering agents is discussed, and the applicability of GLP-1 as a therapeutic
`agent for treatment of type 2 diabetes is highlighted.
`Keywords: GLP-1, diabetes mellitus, insulin secretion.
`
`A. INTRODUCTION
`
`Glucagon-like peptide-1-(7-36)-amide (GLP-1, also
`known as t-GLP-1 or GLIP) is an intestinally-derived
`insulinotropic hormone that has attracted considerable
`attention by virtue of its proven ability to act as a blood
`glucose lowering agent. The efficacy with which GLP-1
`lowers concentrations of blood glucose in type 2 diabetic
`subjects (adult onset diabetes mellitus) has prompted clinical
`investigations whereby the therapeutic value of GLP-1 as an
`antidiabetogenic agent has been substantiated. Earlier studies
`revealed that GLP-1 is an effective stimulator of insulin
`secretion from pancreatic β-cells located in the islets of
`Langerhans. More recently, it has become appreciated that
`GLP-1 also exhibits trophic factor-like properties, acting to
`stimulate β-cell growth and differentiation. These effects of
`GLP-1 are complemented by its ability to suppress appetite
`and to delay gastric emptying. Since GLP-1 is a naturally
`occurring hormone, and because GLP-1 is not reported to
`exert deleterious side effects, new efforts are currently under
`way to develop GLP-1 synthetic analogs that exhibit an
`optimal pharmacokinetic and pharmacodynamic spectrum of
`
`*Address correspondence to this author at the Department of Physiology
`and Neuroscience, MSB 442, New York University School of Medicine,
`550 First Avenue, NY 10016, New York, USA; Tel: 212-263-5434; Fax:
`212-689-9060; E-mail: holzg01@popmail.med.nyu.edu
`
`action commensurate with their use as therapeutic agents.
`The intention of this review is to highlight recent advances
`in this field and to provide an overview of the potential
`usefulness of GLP-1 and its synthetic analogs for treatment
`of type 2 diabetes mellitus. For a detailed discussion of
`previous studies concerning GLP-1, the reader is referred to
`earlier in-depth reviews of this subject matter [1-9].
`
`B. GLP-1 AS A BLOOD GLUCOSE-LOWERING
`AGENT
`
`The blood glucose-lowering hormone GLP-1 possesses
`insulinotropic properties that indicate its usefulness as a
`therapeutic agent in the treatment of diabetes mellitus. When
`administered by intravenous infusion to type 2 diabetic
`subjects, GLP-1 stimulates pancreatic insulin secretion and
`blunts the postprandial hyperglycemic excursion that is
`typically observed following ingestion of a meal [10-12].
`Short term or continuous infusion of GLP-1 restores fasting
`glycemia in type 2 diabetic subjects [13-19], and available
`evidence indicates that GLP-1 remains an effective blood
`glucose lowering agent under conditions in which subjects
`are not responsive to administered sulfonylureas such as
`tolbutamide, glyburide, or glipizide
`[20-22].
`Antidiabetogenic actions of GLP-1 in type 2 diabetic
`subjects are manifest as an improvement of pancreatic β-cell
`function, as measured by a clear restoration of the missing
`
`0929-8673/03 $41.00+.00
`
`© 2003 Bentham Science Publishers Ltd.
`
`MPI EXHIBIT 1031 PAGE 1
`
`MPI EXHIBIT 1031 PAGE 1
`
`

`

`2472 Current Medicinal Chemistry, 2003, Vol. 10, No. 22
`
`Holz and Chepurny
`
`first phase component of glucose-dependent insulin secretion
`[10,15,19,23]. Also observed is an augmentation of second
`phase insulin secretion [14] and an increased amplitude of
`pulsatile insulin secretion [24,25]. Simultaneously, levels of
`fasting blood glucose are reduced (typically by 4-5 mmol/L)
`[19]. Such beneficial actions of administered GLP-1 may be
`a consequence of its ability to compensate for a major deficit
`of endogenous GLP-1 synthesis, secretion, or metabolism in
`diabetic subjects [26,27]. The long term therapeutic value of
`GLP-1 is evident given that it is reported to reduce
`circulating hemoglobin A1c levels by as much as 1.3% [19].
`From the standpoint of its usefulness as a blood glucose
`lowering agent, an attractive pharmacodynamic property of
`GLP-1 is that it stimulates insulin secretion only under
`conditions in which the concentration of blood glucose is
`elevated. As concentrations of blood glucose fall in response
`to administered GLP-1, the insulin secretagogue action of
`GLP-1 is self-terminating [10]. Therefore, unlike
`administered insulin, a natural safeguard exists whereby
`GLP-1 is less likely to induce hypoglycemia in type 2
`diabetic subjects [28,29]. These antidiabetogenic properties
`of GLP-1 are observed not only with intravenous infusion,
`but also
`following subcutaneous administration
`[19,23,29,30-33,] or oral administration via a buccal tablet
`[34].
`Although not fully understood, there is evidence that the
`blood glucose lowering action of GLP-1 is also attributable,
`at least in part, to its ability to suppress pancreatic glucagon
`secretion [11,12,23,35]. This action of the hormone reduces
`hepatic glucose output and may explain, at least in part, an
`ability of GLP-1 to lower levels of blood glucose in type 1
`
`(juvenile-onset) diabetic subjects [11,36]. Interestingly, the
`ability of GLP-1 to suppress pancreatic glucagon secretion
`also appears to be glucose-dependent. Under conditions of
`hypoglycemia, no suppression is observed [37]. Therefore,
`available evidence suggests that GLP-1 does not impair the
`ability of glucagon to act as a counter regulatory hormone in
`support of hepatic glucose output under conditions of
`hypoglycemia.
`It is important to note that GLP-1 is also an inhibitor of
`gastrointestinal secretion and motility, not only in healthy
`individuals [38], but in type 2 diabetic subjects as well
`[21,31,39]. GLP-1 slows gastric emptying and delays
`nutrient absorption, actions that are likely to play a major
`role in determining the effectiveness of GLP-1 as a regulator
`of blood glucose concentration during the time immediately
`following ingestion of a meal [40]. The inhibition of gastric
`emptying by GLP-1 appears to reflect its normal
`physiological role as an intestinal hormone because new
`evidence exists that it mediates the "ileal brake"
`phenomenon [41]. Concomitant with this gastrointestinal
`action, GLP-1 also acts within the central nervous system as
`a mediator of satiety [42]. Evidence has been presented that
`GLP-1 suppresses appetite [43,44], an effect of the hormone
`that appears to involve hypothalamic appetite control centers
`where GLP-1 is synthesized [45,46] and where GLP-1
`receptors are expressed [46,47].
`Although controversial, there is limited evidence that
`GLP-1 exerts an insulinomimetic action to directly facilitate
`glucose uptake in peripheral tissues such as liver, fat or
`muscle. Early on it was recognized that GLP-1 increases
`glucose disposal in type 1 diabetic subjects [11] and healthy
`
`Fig. (1). (A) Alignment of the secretin-like family of peptide hormones including GLP-1-(7-36)-amide. (B) Precursor GLP-1-(1-37)
`aligned with mature forms of the hormone consisting of GLP-1-(7-37) and GLP-1-(7-36)-amide as well as the immediate degradation
`product GLP-1-(9-36)-amide.
`
`MPI EXHIBIT 1031 PAGE 2
`
`MPI EXHIBIT 1031 PAGE 2
`
`

`

`Glucagon-Like Peptide-1 Synthetic Analogs
`
`Current Medicinal Chemistry, 2003, Vol. 10, No. 22 2473
`
`subject [48], while simultaneously reducing the postprandial
`glycemic excursion [35,36,49]. Of particular interest is one
`report that in the OLETF rat model of type 2 diabetes, GLP-
`1 stimulated uptake of 2-deoxy-D-glucose in skeletal muscle
`while exerting no effect on hepatic glucose output [50].
`Therefore, alterations of glucose disposal in response to
`GLP-1 may not be explained simply by a suppression of
`glucagon secretion. This conclusion is supported by a recent
`report in which GLP-1 was shown to increase insulin
`sensitivity in depancreatized dogs [51]. Similarly, GLP-1
`was reported to increase insulin sensitivity and insulin-
`mediated glucose uptake in elderly and obese diabetic
`subjects [52,53]. Despite these observations, no firm
`conclusion can as yet be made concerning the possible
`existence of GLP-1 receptors on hepatocytes, adipocytes, or
`skeletal muscle, and furthermore, such findings have been
`contradicted by several alternative reports [54-58].
`
`C. GLP-1 AND GLP-1 RECEPTOR-MEDIATED
`SIGNAL TRANSDUCTION
`
`In order to appreciate exactly how GLP-1 may exert
`pancreatic and/or extra-pancreatic effects, it is useful to
`review current concepts concerning GLP-1 biosynthesis and
`action. GLP-1 is one member of a large family of
`structurally-related hormones and neuropeptides that include
`glucagon, secretin, glucose-dependent insulinotropic
`polypeptide (GIP), pituitary adenylyl cyclase activating
`polypeptide (PACAP), and vasoactive intestinal polypeptide
`(VIP) (Fig. 1A). GLP-1 is synthesized by enteroendocrine L-
`cells of the distal intestine, and it is released into the
`systemic circulation concomitant with a meal. Secretion is
`stimulated by carbohydrates, fat, hormones, and neural
`reflexes [59-61]. The intestinal L-cells synthesize and
`process proglucagon such that the principal post translational
`end product is GLP-1 rather than glucagon. In vitro analyses
`of GLP-1 biosynthesis have been facilitated by the
`availability of cell lines (GLUTag, NCI-H716) that exhibit
`regulated secretion of the hormone [62-64]. The specificity
`of post translational processing within L-cells distinguishes
`them from pancreatic α-cells where glucagon is the principal
`end product. The immature form of GLP-1 is GLP-1-(1-37),
`and it is thought to be biologically inert (Fig. 1 B) .
`Processing of GLP-1-(1-37) generates the biologically active
`isopeptides GLP-1-(7-37) and GLP-1-(7-36)-amide, with the
`amidated peptide representing the fully mature form of GLP-
`1. Circulating levels of GLP-1 rise to the low picomolar
`range after ingestion of a meal [65], and GLP-1 is rapidly
`inactivated by serum dipeptidyl peptidase IV (DDP-IV; EC
`3.4.14.5) [66-68]. This generates GLP-1-(9-36)-amide, a
`degradation product devoid of agonist activity at the GLP-1
`receptor (Fig. 1B). No evidence exists demonstrating a
`significant difference in pharmacological action when
`comparing GLP-1-(7-37) and GLP-1-(7-36)-amide.
`The GLP-1 receptor cDNA was first identified by use of
`an expression cloning strategy [69]. Subsequently, it was
`established that GLP-1 receptors are expressed in pancreatic
`islets of Langerhans, stomach, lung, heart, kidney, and brain
`[47]. GLP-1 receptors have also been located to vagal
`sensory afferent nerve endings constituting a hepatoportal
`vein glucose sensor [70,71]. This pattern of tissue specific
`
`expression of receptors underlies the ability of GLP-1 to act
`as a stimulus for insulin secretion, to slow gastric emptying,
`and to suppress appetite. The GLP-1 receptor genomic
`sequence identifies it as a member of Class II (Family B)
`heptahelical GPCRs. Receptors of this family are
`structurally-related and recognize GLP-1, glucagon, GIP,
`secretin, PACAP, VIP, calcitonin, parathyroid hormone
`(PTH), and corticotropin releasing factor (CRF).
`A common feature of Class II GPCRs is their ability to
`mediate stimulatory actions of peptide hormones on cAMP
`production, Ca2+ signaling, and exocytosis. This is also true
`for GLP-1 receptors expressed on pancreatic β-cells. By
`stimulating production of cAMP, GLP-1 acts as a modulator
`of the β-cell glucose signaling system (Fig.2). The GLP-1
`receptor is positively coupled to adenylyl cyclase by
`heterotrimeric Gs proteins [72]. Evidence also exists for
`coupling of GLP-1 receptors to Gi and Gq/11 proteins [73].
`GLP-1 receptor occupancy stimulates cAMP production,
`whereas very little evidence exists for a major effect of GLP-
`1 on β-cell inositol phosphate production. PKA is a down
`stream effector of the GLP-1 receptor, and it is activated by
`an increase of [cAMP]i. Evidence exists for the targeting of
`PKA to specific subcellular compartments via A-kinase
`anchoring proteins (AKAPs) [74,75], thereby offering one
`potential explanation for how the cAMP-dependent actions
`of GLP-1 may be spatially restricted within the β-cell. An
`additional target of cAMP action is the newly recognized
`family of cAMP-binding proteins known as cAMP-regulated
`guanine nucleotide exchange factors (cAMPGEFs, also
`referred to as Epac) [76-79]. The cAMPGEFs couple cAMP
`production to the activation of Rap1, a small molecular
`weight G protein. Novel signaling properties of the GLP-1
`receptor include its ability to activate immediate early genes
`(IEGs) [80], to increase the number of insulin receptors on
`insulin-secreting cells [81,82], to stimulate mitogen-
`activated protein kinases
`(p38 MAPK, ERK),
`phosphatidylinositol 3-kinase [83], atypical protein kinase
`C-ζ [84], Ca2+/calmodulin-regulated protein kinase [85],
`protein kinase B (Akt), and hormone-sensitive lipase [86]. It
`seems likely that at least some of these effects result from
`activation of Epac by GLP-1. Available evidence also
`indicates an important action of GLP-1 to regulate β-cell ion
`channel function. GLP-1 has been reported to inhibit ATP-
`sensitive K+ channels (K-ATP) [87-89] and to facilitate the
`opening of voltage-dependent Ca2+ channels (VDCCs) [3].
`Intracellular Ca2+ release channels are also targeted by GLP-
`1 in a cAMP-dependent manner [74,90,91]. They include the
`type-2 isoform of ryanodine receptor (RYR-2) Ca2+ release
`channels located on the endoplasmic reticulum.
`
`D. GLP-1 AS A ββββ-CELL GLUCOSE COMPETENCE
`HORMONE
`
`It is now established that GLP-1 acts a competence
`hormone in support of glucose-dependent insulin secretion
`[1,87,92,93]. Under conditions in which β-cells are
`metabolically compromised, GLP-1 acts to restore the
`sensitivity of these cells to glucose. Since type 2 diabetes
`mellitus is a metabolic disorder in which β-cells lose their
`ability to respond to glucose, the induction of glucose
`competence by GLP-1 may be of major therapeutic
`
`MPI EXHIBIT 1031 PAGE 3
`
`MPI EXHIBIT 1031 PAGE 3
`
`

`

`2474 Current Medicinal Chemistry, 2003, Vol. 10, No. 22
`
`Holz and Chepurny
`
`Fig. (2). Illustrated is the role GLP-1 plays as a modulator of the β-cell glucose signaling system. Glucose stimulates insulin
`secretion, and GLP-1 potentiates the action of glucose by activating multiple signal transduction pathways important to Ca2+-
`dependent exocytosis. Uptake of glucose is mediated by the type-2 glucose transporter (Glut2) and glucose is converted to glucose-6
`phosphate by glucokinase, a type-IV hexokinase that is rate-limiting for β-cell glucose sensing. Aerobic glycolysis generates
`metabolic coupling factors, one of which is ATP. An increase of the cytosolic ATP/ADP concentration ratio results in the closing of
`ATP-sensitive K+ channels (K-ATP), thereby producing membrane depolarization and activation of Ca2+ influx through voltage-
`sensitive Ca2+ channels (Ca-VS). Ca2+ influx produces an increase of [Ca2+]i and triggers fusion of insulin secretory granules with the
`plasma membrane. Repolarization of the membrane is due, in part, to the Ca2+-dependent activation of K+ channels (K-Ca). GLP-1
`modulates this sequence of events via second messengers (cAMP, Ca2+), protein kinase A (PKA), guanine nucleotide exchange factors
`(Epac), Ca2+-calmodulin-regulated protein kinase (CaMK), hormone sensitive lipase (HSL), and lipid metabolites including free fatty
`acids (FFAs).
`
`importance. Recent studies suggest that glucose competence
`results from the ability of GLP-1 to act as a glucose-
`sensitizer. The central locus for this effect appears to be
`intermediary metabolism where GLP-1 facilitates glucose-
`dependent mitochondrial ATP production [94]. This key
`observation provides a clear explanation for why the insulin
`secretagogue action of GLP-1 in β-cells is entirely glucose-
`dependent. An initiating event for induction of glucose
`competence is likely to be the release of Ca2+ from
`intracellular Ca2+ stores. This is Ca2+-induced Ca2+ release
`(CICR), and it is stimulated by GLP-1. Available evidence
`suggests that endoplasmic reticulum-derived Ca2+ interacts
`with cAMP
`to facilitate mitochondrial oxidative
`phosphorylation. Intramitochondrial targets of Ca2+ include
`Ca2+-sensitive dehydrogenases, whereas the targets of cAMP
`may include mitochondrial PKA and Epac, although this
`remains to be determined. Regardless of the precise signal
`transduction mechanism involved, it is reasonable to
`hypothesize that the induction of β-cell glucose competence
`by GLP-1 might contribute to its ability to lower
`concentrations of blood glucose in type 2 diabetic subjects.
`
`Indeed, the restoration of first phase insulin secretion by
`GLP-1 in diabetic subjects is understandable in view of this
`hormone's ability to augment glucose-dependent ATP
`production, thereby closing β-cell K-ATP channels. This
`action of GLP-1 at K-ATP is analogous to the effect
`produced by sulfonylureas (tolbutamide, glyburide,
`glipizide), however it is unique in that it is entirely
`dependent on metabolism of glucose rather than being the
`consequence of a direct pharmacological inhibition of K-
`ATP.
`
`E. THE INSULIN SECRETAGOGUE ACTION OF
`GLP-1
`
`GLP-1 stimulates pancreatic insulin secretion under
`conditions in which β-cells are exposed to concentrations of
`glucose typical of the postprandial state (> 7.5 mM). Insulin
`secretion occurs in a pulsatile manner, and GLP-1 increases
`the amplitude of each pulse without changing the pulse
`frequency [24,25,95]. These effects of GLP-1 are
`accompanied by increased oscillatory electrical activity of the
`
`MPI EXHIBIT 1031 PAGE 4
`
`MPI EXHIBIT 1031 PAGE 4
`
`

`

`Glucagon-Like Peptide-1 Synthetic Analogs
`
`Current Medicinal Chemistry, 2003, Vol. 10, No. 22 2475
`
`islets due to the generation of action potentials [96].
`Evidence exists indicating that GLP-1 facilitates both the
`triggering (first phase) and augmentation (second phase)
`pathways of glucose-dependent insulin secretion [3].
`Stimulatory effects of GLP-1 on insulin secretion appear to
`be mediated primarily by cAMP [3]. New findings indicate
`that GLP-1 and cAMP target insulin granule-associated
`proteins including Epac [76] and Rim2 (77), thereby
`increasing the likelihood that a readily releasable pool of
`secretory granules will undergo exocytosis in response to an
`increase of intracellular Ca2+ concentration. Simultaneously,
`GLP-1 appears to stimulate refilling of the readily releasable
`pool, an effect attributed to the mobilization of a reserve
`pool of secretory granules [97]. Such direct effects of GLP-1
`on the exocytotic secretory apparatus are complimented by
`the ability of GLP-1 to increase β-cell electrical activity,
`thereby facilitating influx of Ca2+ through VDCCs [87].
`Simultaneously, GLP-1 mobilizes Ca2+ from intracellular
`Ca2+ stores via CICR, thereby amplifying the exocytosis
`triggered by Ca2+ influx [98]. The Ca2+ stores mobilized by
`GLP-1 appear to correspond to those that are sensitive to
`ryanodine, caffeine, and thapsigargin [75,98]. Indeed,
`evidence has been presented that β-cell RYR Ca2+ release
`channels are important intermediaries linking GLP-1-
`stimulated cAMP production to Ca2+ mobilization and the
`initiation of Ca2+-dependent insulin granule exocytosis [98].
`
`STIMULATION OF
`F.
`EXPRESSION BY GLP-1
`
`INSULIN GENE
`
`GLP-1 stimulates insulin gene expression by virtue of its
`ability to increase transcription of the insulin gene while
`simultaneously stabilizing preproinsulin mRNA [99-103].
`GLP-1 also increases translational biosynthesis of
`proinsulin. These effects of GLP-1 resemble the previously
`described action of glucose to increase β-cell insulin content.
`The ability of GLP-1 to stimulate insulin gene transcription
`has been studied in isolated islets of Langerhans and in a
`variety of insulinoma cell lines. In general, GLP-1 exerts
`multiple stimulatory influences on insulin gene promoter
`activity, as expected given that it activates more than one
`signal transduction pathway. These signaling pathways
`converge at the promoter to regulate the function of
`
`transcription factors that interact with specific response
`elements (Fig.3). Although a conventional cAMP signaling
`mechanism involving PKA, CREB, and the insulin gene
`cAMP response element (CRE) has been suggested to play a
`significant role in this effect, it now appears more likely that
`actions of GLP-1 at the CRE are PKA-independent. This
`conclusion is based on studies of the INS-1 insulin-secreting
`cell line where it was demonstrated that pharmacological
`inhibitors of PKA (H-89, KT 5720) failed to block
`stimulatory actions of GLP-1 at a luciferase reporter
`incorporating -410 bp of the rat insulin I gene promoter
`(RIP1-Luc). Interestingly, the action of GLP-1 was shown to
`be blocked by the serine/threonine protein kinase inhibitor
`Ro 31-8220, by transfection of INS-1 cells with a dominant
`negative isoform of CREB (A-CREB), or by introduction of
`inactivating mutations at the CRE. On the basis of these
`observations, it was suggested that stimulatory actions of
`GLP-1 at the CRE are mediated by basic region leucine
`zipper (bZIP) transcription factor related in structure to
`CREB, and that the transactivation function of such bZIPs
`might be upregulated by an Ro 31-8220-sensitive MAPK-
`activated kinase such as RSK and/or MSK.
`the
`that
`Evidence has also been presented
`pancreatic/duodenal homeodomain transcription factor PDX-
`1 mediates stimulatory actions of GLP-1 at A-elements of
`the insulin gene promoter. PDX-1 translocates to the nucleus
`in response to GLP-1, an effect mediated by PKA [104].
`Levels of PDX-1 mRNA are increased by GLP-1 [105,106],
`and binding of PDX-1 to A1 elements of the rat insulin I
`and II gene promoters is facilitated. The transactivation
`function of PDX-1 is stimulated by GLP-1 [107], and GLP-
`1 also stimulates a luciferase reporter incorporating synthetic
`multimerized E2/A4/A3 elements of RIP1. Given that the
`A-elements of the insulin gene promoter are established to
`be mediators of glucose insulinotropic action, they appear to
`be a locus at which nutrient metabolism and hormonal
`signal transduction interact.
`Recently, it has been suggested that NFAT transcription
`factors (nuclear factor of activated T-cells) mediate
`stimulatory effects of GLP-1 on insulin gene transcription
`[108]. Three NFAT binding sites have been identified in the
`rat insulin I gene promoter (RIP1), and it was suggested that
`GLP-1 facilitates binding of NFAT to the promoter in a
`
`Fig. (3). Response elements and transcription factors that are targeted by GLP-1 for increased insulin gene transcription. Within the
`first -400 bp of the promoter are found 3 predicted response elements for NFAT (NFAT-RE-1-3). Also present are A4/A3 elements that
`bind the homeodomain transcription factor PDX-1. A non-palindromic cyclic AMP response element (CRE) mediates stimulatory
`effects of GLP-1 via basic region leucine zipper (bZIP) transcription factors related in structure to CREB.
`
`MPI EXHIBIT 1031 PAGE 5
`
`MPI EXHIBIT 1031 PAGE 5
`
`

`

`2476 Current Medicinal Chemistry, 2003, Vol. 10, No. 22
`
`Holz and Chepurny
`
`cAMP/PKA and Ca2+-dependent manner. Analogous to the
`previously described stimulatory effect of cAMP and Ca2+ at
`the glucagon gene promoter [109], it was suggested that the
`cAMP and Ca2+-dependence of GLP-1 action at RIP1 is
`explained by activation of protein phosphatase 2B (PP2B,
`calcineurin). In this scheme, PP2B-mediated dephosphoryla-
`tion of NFAT leads to its nuclear translocation and
`subsequent stimulation of insulin gene transcription.
`
`G. NEWLY RECOGNIZED GROWTH FACTOR-
`LIKE EFFECTS OF GLP-1
`Substantial evidence now exists that GLP-1 acts as a β-
`cell growth factor with neogenic [106,110], mitogenic
`[83,84,111,112] and anti-apoptotic activities [112-114].
`Administration of GLP-1 or the GLP-1 analog Exendin-4 to
`rats increases β-cell mass in a partial pancreatectomy model
`of type 2 diabetes [110]. This effect has also been observed
`in rats using the streptozotocin model of diabetes [115], the
`Goto-Kakizaki model of genetic diabetes [116], or in
`glucose-intolerant rats of advanced age [117]. Similarly,
`Exendin-4 and the long lasting GLP-1 derivative NN2211
`increase β-cell mass in db/db diabetic mice [112,118]. In
`each of these animal models it has been suggested that the
`increase of β -cell mass results from the increased
`differentiation of β-cells from precursor pancreatic ductal
`stem cells (neogenesis). Surprisingly, the proliferative
`capacity of preexisting β-cells appears to be stimulated,
`thereby explaining the mitogenic action of GLP-1. These
`effects of GLP-1 and Exendin-4 are associated with increased
`expression of PDX-1, and in fact, the stimulation of PDX-1
`expression may be a pivotal event underlying the
`differentiation of ductal stem cells to β-cells [106,117-123].
`Neogenesis in response to GLP-1 or Exendin-4 has been
`studied in vitro using primary cultures of progenitor β-cells.
`Evidence was presented that GLP-1 and Exendin-4 induce
`the differentiation of human pancreatic islet-derived
`progenitor cells into insulin-producing cells [122,124].
`Findings similar to these were also obtained using primary
`cultures of rat pancreatic ductal cells [125]. Recently, it has
`been suggested that GLP-1 acts not only as a growth factor
`in support of β-cell neogenesis, but also as a morphogen for
`embryonic development of the pancreas [124,126]. It may be
`concluded that the in vivo therapeutic actions of GLP-1 may
`include its ability to stimulate pancreatic growth and
`differentiation in human diabetic subjects where β-cell mass
`is insufficient to compensate for insulin resistance.
`The increase of β-cell mass observed in response to long
`term (days) treatment with GLP-1 is preceded by a short
`term (hrs) action of the hormone. This short term effect is
`referred to as "functional maturation" whereby immature β-
`cells become competent to secrete insulin in response to
`glucose. Although functional maturation has been described
`in studies of fetal human and pig islet-like cell clusters
`[127,128], it is not clear at the present time whether this
`phenomenon results from immediate (i.e., within seconds)
`effects of GLP-1 on intermediary metabolism (as is the case
`for the induction of Glucose Competence), or alternatively,
`major alterations in the level of expression of key signaling
`molecules subserving β-cell glucose signaling (Glut2
`transporter, glucokinase, K-ATP channels).
`
`To investigate the mitogenic activity of GLP-1, Buteau
`and co-workers [129] studied INS-1 insulin-secreting cells
`that serve as a model system for analyses of β-cell signal
`transduction. It was reported that the proliferation of INS-1
`cells in response to GLP-1 results from "transactivation" of
`the EGF receptor. In this signaling event, GLP-1 activates c-
`Src and promotes the release of a soluble EGF receptor
`ligand (betacelluline), thereby initiating a cascade of protein
`kinase-mediated phosphorylation reactions catalyzed by PI-
`3K, atypical PKC-ζ, and PKB, among others [112,129].
`These findings are of interest because they suggest a possible
`usefulness of GLP-1 as a stimulus for the in vitro expansion
`of progenitor β-cells prior to induction of differentiation.
`Such a strategy might allow large numbers of β-cells to be
`derived in cell cultures, thereby providing a source of
`insulin-secreting cells for use in transplantation.
`The beneficial action of GLP-1 extends to its ability to
`slow β-cell apoptosis and to protect against stress. For
`example, GLP-1 and Exendin-4 protect against β-cell
`apoptosis resulting from the treatment of mice with
`streptozotocin [114]. Similarly, both GLP-1 and Exendin-4
`protect against cytokine-induced cell death in preparations of
`rat β-cells [114]. The importance of such findings is
`emphasized by a recent report demonstrating that the
`protective action of GLP-1 is evident in Zucker diabetic
`fa/fa rats, a genetic model of obesity-related type 2 diabetes
`[113]. It is also of interest that GLP-1 appears to exert a
`cytoprotective action not only in β-cells, but also in neurons
`[130]. Such findings extend on previous studies
`demonstrating that PACAP, a neuropeptide related in
`structure to GLP-1, exerts a neuroprotective effect within the
`brain [131]. On the basis of these observations, it may be
`speculated that one unexpected and beneficial therapeutic
`action of GLP-1 may be its ability to protect against β-cell
`apoptosis associated with autoimmune destruction (type 1
`diabetes) or β-cell exhaustion (type 2 diabetes).
`In conclusion, the above summarized actions of GLP-1
`are understandable if there exists a compensatory mechanism
`whereby biosynthesis of GLP-1 is stimulated under
`conditions in which β-cells are stressed. Evidence that this is
`in fact the case is provided by one recent report
`demonstrating that the biosynthesis of GLP-1 is stimulated
`in rats made diabetic by treatment with streptozotocin [132].
`In this animal model of diabetes, increased biosynthesis of
`GLP-1 occurs secondary to the increased expression of a
`prohormone converting enzyme (PC1/3) that processes
`proglucagon to generate GLP-1. Evidently, accelerated
`biosynthesis of GLP-1 is an evolutionary adaptation to
`stress, thereby allowing an organism to match intra-islet
`levels of GLP-1 to the prevailing need for β - c e l l
`mitogenesis, neogenesis, and differentiation. Summarized in
`(Fig.4) are the multiple signaling pathways by which GLP-1
`influences growth, differentiation, neogenesis, survival,
`insulin biosynthesis, and insulin secretion in β-cells.
`
`H. GLP-1 SYNTHETIC ANALOGS OBTAINED BY
`CHEMICAL MODIFICATION
`
`Clinical studies have demonstrated that the blood
`glucose lowering action of GLP-1 is transient owing to the
`short plasma half life (1.5-5.0 min) of the peptide. Sustained
`
`MPI EXHIBIT 1031 PAGE 6
`
`MPI EXHIBIT 1031 PAGE 6
`
`

`

`Glucagon-Like Peptide-1 Synthetic Analogs
`
`Current Medicinal Chemistry, 2003, Vol. 10, No. 22 2477
`
`Fig. (4). Summary of the biological actions of GLP-1 in β-cells. Illustrated are the second messengers, kinases, and transcription
`factors that mediate insulinotropic effects of GLP-1 on insulin biosynthesis and secretion. Also illustrated are the growth factor-like
`actions of GLP-1 that are important to proliferation, neogenesis, differentiation, and survival of β-cells.
`
`lowering of blood glucose concentration is only observed
`with continuous infusion, as demonstrated in studies in
`which GLP-1 was administered by intravenous infusion over
`a 24 hr time course [22]. Clearly, this mode of
`administration is not compatible with general use by the
`public. To address this short coming, efforts have been
`directed to engineer long lasting analogs of GLP-1 that are
`synthetic peptides and which exhibit insulinotropic and
`blood glucose lowering activities in vivo. These peptides
`include NN221

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket